Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 525
Filtrar
1.
J Immunol Methods ; 526: 113618, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38237697

RESUMO

The high burden of disease and the long-lasting sequelae following Streptococcus pyogenes (Strep A) infections make the development of an effective vaccine a global health priority. Streptolysin O (SLO), is a key toxin in the complex pathogenesis of Strep A infection. Antibodies are elicited against SLO after natural exposure and represent a key target for vaccine-induced immunity. Here we present the setup and characterization of a hemolysis assay to measure functionality of anti-SLO antibodies in human sera. Assay specificity, precision, linearity, reproducibility, and repeatability were determined. The assay was demonstrated to be highly sensitive, specific, reproducible, linear and performed well in assessing functionality of anti-SLO antibodies induced by exposed individuals. Moreover, different sources of critical reagents, in particular red- blood cells, have been compared and had minimal impact on assay performance. The assay presented here has throughput suitable for evaluating sera in vaccine clinical trials and sero-epidemiological studies to gain further insights into the functionality of infection- and vaccine-induced antibodies.


Assuntos
Infecções Estreptocócicas , Vacinas , Humanos , Streptococcus pyogenes , Hemólise , Reprodutibilidade dos Testes , Estreptolisinas/farmacologia , Proteínas de Bactérias , Anticorpos/farmacologia , Infecções Estreptocócicas/diagnóstico
2.
Molecules ; 27(19)2022 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-36234795

RESUMO

Streptococcus pneumoniae (S. pneumoniae), as a Gram-positive bacterium, can cause severe bacterial pneumonia, and result in high morbidity and mortality in infected people. Meanwhile, isolated drug-resistant S. pneumoniae is growing, which raises concerns about strategies for combatting S. pneumoniae infection. To disturb S. pneumoniae pathogenicity and its drug-resistance, developing novel anti-infective strategies or compounds is urgent. In this study, the anti-infective effect of shionone was explored. A minimum inhibitory concentration (MIC) assay and growth curve determination were performed to evaluate the effect of the tetracyclic triterpenoid compound shionone against S. pneumoniae. Hemolysis tests, western blotting, oligomerization inhibition assays, and molecular docking were carried out to explore the anti-infective mechanism of shionone. Moreover, the protective effect of shionone was also confirmed in a mousepneumonia model. The results showed that the excellent hemolytic inhibitory activity of shionone was observed at less than 8 µg/mL. Meanwhile, shionone could disturb the oligomerization of pneumolysin (PLY) but did not interfere with PLY expression at less than 4 µg/mL. Molecular docking suggested that shionone targeted the ASP-59, ILE-60, THR-57, PHE-344, and ASN-346 amino acid sites to reduce S. pneumoniae pathogenicity. Furthermore, shionone alleviated lung histopathologic injury and decreased lung bacterial colonization in vivo. The above results showed that shionone could bind to the PLY active pocket under the concentrations of 8 µg/mL and neutralize PLY hemolysis activity to reduce S. pneumoniae pathogenicity in vitro and in vivo.


Assuntos
Lesão Pulmonar Aguda , Triterpenos , Aminoácidos/farmacologia , Proteínas de Bactérias/metabolismo , Hemólise , Humanos , Simulação de Acoplamento Molecular , Streptococcus pneumoniae , Estreptolisinas/metabolismo , Estreptolisinas/farmacologia , Triterpenos/farmacologia
3.
Molecules ; 27(16)2022 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-36014299

RESUMO

Pneumolysin (PLY) is a significant virulence factor of Streptococcus pneumoniae (S. pneumoniae), able to break through the defense system of a host and mediate the occurrence of a series of infections. Therefore, PLY as the most ideal target to prevent S. pneumoniae infection has received more and more attention and research. Corilagin is a tannic acid that exhibits excellent inhibition of PLY oligomers without bacteriostatic activity to S. pneumoniae. Herein, hemolytic activity assays, cell viability tests and western blot experiments are executed to evaluate the antivirulence efficacy of corilagin against PLY in vitro. Colony observation, hematoxylin and eosin (H&E) staining and cytokines of bronchoalveolar lavage fluid (BALF) are applied to assess the therapeutic effect of corilagin in mice infected by S. pneumoniae. The results indicate the related genes of corilagin act mainly via enrichment in pathways associated with pneumonia disease. Furthermore, molecular docking and molecular dynamics simulations show that corilagin might bind with domains 3 and 4 of PLY and interfere with its hemolytic activity, which is further confirmed by the site-directed mutagenesis of PLY. Additionally, corilagin limits PLY oligomer production without impacting PLY expression in S. pneumoniae cultures. Moreover, corilagin effectively relieves PLY-mediated cell injury without any cytotoxicity, even then reducing the colony count in the lung and the levels of pro-inflammatory factors in BALF and remarkably improving lung lesions. All the results demonstrate that corilagin may be a novel strategy to cope with S. pneumoniae infection by inhibiting PLY oligomerization.


Assuntos
Infecções Pneumocócicas , Estreptolisinas , Animais , Proteínas de Bactérias/metabolismo , Glucosídeos , Hemólise , Taninos Hidrolisáveis , Camundongos , Simulação de Acoplamento Molecular , Infecções Pneumocócicas/tratamento farmacológico , Streptococcus pneumoniae , Estreptolisinas/metabolismo , Estreptolisinas/farmacologia
4.
Immunology ; 167(3): 413-427, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35835695

RESUMO

Published data for the Streptococcus pneumoniae virulence factor Pneumolysin (Ply) show contradictory effects on the host inflammatory response to infection. Ply has been shown to activate the inflammasome, but also can bind to MRC-1 resulting in suppression of dendritic cell inflammatory responses. We have used an in vitro infection model of human monocyte-derived macrophages (MDM), and a mouse model of pneumonia to clarify whether pro- or anti-inflammatory effects dominate the effects of Ply on the initial macrophage inflammatory response to S. pneumoniae, and the consequences during early lung infection. We found that infection with S. pneumoniae expressing Ply suppressed tumour necrosis factor (TNF) and interleukin-6 production by MDMs compared to cells infected with ply-deficient S. pneumoniae. This effect was independent of bacterial effects on cell death. Transcriptional analysis demonstrated S. pneumoniae expressing Ply caused a qualitatively similar but quantitatively lower MDM transcriptional response to S. pneumoniae compared to ply-deficient S. pneumoniae, with reduced expression of TNF and type I IFN inducible genes. Reduction of the MDM inflammatory response was prevented by inhibition of SOCS1. In the early lung infection mouse model, the TNF response to ply-deficient S. pneumoniae was enhanced and bacterial clearance increased compared to infection with wild-type S. pneumoniae. Overall, these data show Ply inhibits the initial macrophage inflammatory response to S. pneumoniae, probably mediated through SOCS1, and this was associated with improved immune evasion during early lung infection.


Assuntos
Inflamassomos , Streptococcus pneumoniae , Animais , Anti-Inflamatórios , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Interleucina-6 , Macrófagos/metabolismo , Camundongos , Estreptolisinas/genética , Estreptolisinas/metabolismo , Estreptolisinas/farmacologia , Fatores de Necrose Tumoral , Fatores de Virulência
5.
Cells ; 10(12)2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34944089

RESUMO

Pneumolysin (PLY) is a pore-forming toxin of Streptococcus pneumoniae that contributes substantially to the inflammatory processes underlying pneumococcal pneumonia and lung injury. Host responses against S. pneumoniae are regulated in part by neutrophils and platelets, both individually and in cooperative interaction. Previous studies have shown that PLY can target both neutrophils and platelets, however, the mechanisms by which PLY directly affects these cells and alters their interactions are not completely understood. In this study, we characterize the effects of PLY on neutrophils and platelets and explore the mechanisms by which PLY may induce neutrophil-platelet interactions. In vitro studies demonstrated that PLY causes the formation of neutrophil extracellular traps (NETs) and the release of extracellular vesicles (EVs) from both human and murine neutrophils. In vivo, neutrophil EV (nEV) levels were increased in mice infected with S. pneumoniae. In platelets, treatment with PLY induced the cell surface expression of P-selectin (CD62P) and binding to annexin V and caused a significant release of platelet EVs (pl-EVs). Moreover, PLY-induced nEVs but not NETs promoted platelet activation. The pretreatment of nEVs with proteinase K inhibited platelet activation, indicating that the surface proteins of nEVs play a role in this process. Our findings demonstrate that PLY activates neutrophils and platelets to release EVs and support an important role for neutrophil EVs in modulating platelet functions in pneumococcal infections.


Assuntos
Vesículas Extracelulares/metabolismo , Neutrófilos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Estreptolisinas/farmacologia , Animais , Proteínas de Bactérias/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Armadilhas Extracelulares/efeitos dos fármacos , Armadilhas Extracelulares/metabolismo , Humanos , Camundongos , Ativação de Neutrófilo/efeitos dos fármacos
6.
Parasitol Res ; 119(12): 4297-4302, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33089360

RESUMO

Malaria is caused by unicellular parasites of the genus Plasmodium, which reside in erythrocytes during the clinically relevant stage of infection. To separate parasite from host cell material, haemolytic agents such as saponin are widely used. Previous electron microscopy studies on saponin-treated parasites reported both, parasites enclosed by the erythrocyte membrane and liberated from the host cell. These ambiguous reports prompted us to investigate haemolysis by live-cell time-lapse microscopy. Using either saponin or streptolysin O to lyse Plasmodium falciparum-infected erythrocytes, we found that ring-stage parasites efficiently exit the erythrocyte upon haemolysis. For late-stage parasites, we found that only approximately half were freed, supporting the previous electron microscopy studies. Immunofluorescence imaging indicated that freed parasites were surrounded by the parasitophorous vacuolar membrane. These results may be of interest for future work using haemolytic agents to enrich for parasite material.


Assuntos
Eritrócitos/parasitologia , Hemólise/efeitos dos fármacos , Plasmodium falciparum/fisiologia , Saponinas/farmacologia , Estreptolisinas/farmacologia , Proteínas de Bactérias/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/parasitologia , Eritrócitos/efeitos dos fármacos , Vesículas Extracelulares/parasitologia , Humanos , Estágios do Ciclo de Vida/fisiologia , Microscopia , Plasmodium falciparum/crescimento & desenvolvimento
7.
Nat Commun ; 11(1): 5018, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33024089

RESUMO

The re-emergence of scarlet fever poses a new global public health threat. The capacity of North-East Asian serotype M12 (emm12) Streptococcus pyogenes (group A Streptococcus, GAS) to cause scarlet fever has been linked epidemiologically to the presence of novel prophages, including prophage ΦHKU.vir encoding the secreted superantigens SSA and SpeC and the DNase Spd1. Here, we report the molecular characterization of ΦHKU.vir-encoded exotoxins. We demonstrate that streptolysin O (SLO)-induced glutathione efflux from host cellular stores is a previously unappreciated GAS virulence mechanism that promotes SSA release and activity, representing the first description of a thiol-activated bacterial superantigen. Spd1 is required for resistance to neutrophil killing. Investigating single, double and triple isogenic knockout mutants of the ΦHKU.vir-encoded exotoxins, we find that SpeC and Spd1 act synergistically to facilitate nasopharyngeal colonization in a mouse model. These results offer insight into the pathogenesis of scarlet fever-causing GAS mediated by prophage ΦHKU.vir exotoxins.


Assuntos
Exotoxinas/metabolismo , Prófagos/genética , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/virologia , Animais , Proteínas de Bactérias/farmacologia , Linhagem Celular , Eritrócitos/efeitos dos fármacos , Exotoxinas/genética , Feminino , Glutationa/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Faringe/citologia , Escarlatina/epidemiologia , Escarlatina/microbiologia , Streptococcus pyogenes/genética , Estreptolisinas/farmacologia , Superantígenos/genética , Superantígenos/metabolismo
8.
Cells ; 9(5)2020 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397494

RESUMO

Microbial proteins have recently been found to have more benefits in clinical disease treatment because of their better-developed strategy and properties than traditional medicine. In this study, we investigated the effectiveness of a truncated peptide synthesized from the C-terminal sequence of pneumolysin, i.e., C70PLY4, in Streptococcus pneumoniae, in treating chronic inflammatory conditions. It has been shown that C70PLY4 significantly blocks the transendothelial migration of neutrophils and attenuates the formation of atherosclerotic plaque and the secretion of soluble forms of the intercellular adhesion molecule-1 (ICAM-1), the vascular cell adhesion molecule 1 (VCAM-1), and E-selectin in high-fat-diet/streptozotocin-induced inflammatory rats. The mechanism and the docking simulation analysis further indicated that C70PLY4 might serve as a Toll-like receptor 4 (TLR4) antagonist by competing for the binding site of MD2, an indispensable protein for lipopolysaccharide (LPS)-TLR4 interaction signaling, on the TLR4 structure. Moreover, compared to the full-length PLY, C70PLY4 seems to have no cytotoxicity in human vascular endothelial cells. Our study elucidated a possible therapeutic efficacy of C70PLY4 in reducing chronic inflammatory conditions and clarified the underlying mechanism. Thus, our findings identify a new drug candidate that, by blocking TLR4 activity, could be an effective treatment for patients with chronic inflammatory diseases.


Assuntos
Inflamação/tratamento farmacológico , Proteínas Mutantes/farmacologia , Proteínas Mutantes/uso terapêutico , Streptococcus pneumoniae/metabolismo , Estreptolisinas/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Sítios de Ligação , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dieta Hiperlipídica , Selectina E/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos , Camundongos , Simulação de Acoplamento Molecular , Proteínas Mutantes/química , NF-kappa B/metabolismo , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Solubilidade , Estreptolisinas/química , Estreptozocina , Receptor 4 Toll-Like/metabolismo , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
Methods Mol Biol ; 2136: 367-375, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32430837

RESUMO

Group A Streptococcus (GAS) produces the pore-forming toxin, streptolysin O (SLO). SLO sequesters cholesterol and induces a plasma membrane repair process that removes the pores via a lipid raft-mediated endocytosis. The impact SLO has on membranes makes it an effective toxin for investigating the function of lipid rafts in cellular processes. Lipid rafts are essential for B-cell activation. Indeed, antigen-stimulated B-cell receptors (BCRs) require localization with lipid rafts for efficient signaling and internalization. SLO treatment impairs BCR activation by competing for lipid rafts. Here, disrupting lipid rafts using SLO and assessing the effects on BCR activation by fluorescence microscopy and flow cytometry are described.


Assuntos
Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/fisiologia , Estreptolisinas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Membrana Celular/metabolismo , Colesterol/metabolismo , Endocitose , Ativação Linfocitária , Microdomínios da Membrana/efeitos dos fármacos , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/fisiologia , Streptococcus pyogenes/metabolismo , Estreptolisinas/farmacologia
10.
Am J Physiol Cell Physiol ; 318(6): C1226-C1237, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32348180

RESUMO

The ubiquitous calpains, calpain-1 and -2, play important roles in Ca2+-dependent membrane repair. Mechanically active tissues like skeletal muscle are particularly reliant on mechanisms to repair and remodel membrane injury, such as those caused by eccentric damage. We demonstrate that calpain-1 and -2 are master effectors of Ca2+-dependent repair of mechanical plasma membrane scrape injuries, although they are dispensable for repair/removal of small wounds caused by pore-forming agents. Using CRISPR gene-edited human embryonic kidney 293 (HEK293) cell lines, we established that loss of both calpains-1 and -2 (CAPNS1-/-) virtually ablates Ca2+-dependent repair of mechanical scrape injuries but does not affect injury or recovery from perforation by streptolysin-O or saponin. In contrast, cells with targeted knockout of either calpain-1 (CAPN1-/-) or -2 (CAPN2-/-) show near-normal repair of mechanical injuries, inferring that both calpain-1 and calpain-2 are equally capable of conducting the cascade of proteolytic cleavage events to reseal a membrane injury, including that of the known membrane repair agent dysferlin. A severe muscular dystrophy in a murine model with skeletal muscle knockout of Capns1 highlights vital roles for calpain-1 and/or -2 for health and viability of skeletal muscles not compensated for by calpain-3 (CAPN3). We propose that the dystrophic phenotype relates to loss of maintenance of plasma membrane/cytoskeletal networks by calpains-1 and -2 in response to directed and dysfunctional Ca2+-signaling, pathways hyperstimulated in the context of membrane injury. With CAPN1 variants associated with spastic paraplegia, a severe dystrophy observed with muscle-specific loss of calpain-1 and -2 activity identifies CAPN2 and CAPNS1 as plausible candidate neuromuscular disease genes.


Assuntos
Calpaína/deficiência , Membrana Celular/enzimologia , Músculo Esquelético/enzimologia , Distrofia Muscular do Cíngulo dos Membros/enzimologia , Distrofia Muscular Animal/enzimologia , Animais , Proteínas de Bactérias/farmacologia , Sinalização do Cálcio , Calpaína/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/patologia , Modelos Animais de Doenças , Disferlina/deficiência , Disferlina/genética , Feminino , Células HEK293 , Humanos , Masculino , Camundongos Knockout , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Saponinas/farmacologia , Índice de Gravidade de Doença , Estreptolisinas/farmacologia
11.
Biochem Biophys Res Commun ; 517(4): 596-602, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31395343

RESUMO

The pneumolysin (Ply) protein of Streptococcus pneumoniae is composed of four domains and possesses several different but related activities. In this study, recombinant Ply and two truncated forms, Ply domain 1-3 and Ply domain 4 (rPly4), were expressed and characterized regarding their participation in apoptosis, the stimulation of cytokine production, hemolytic activity and virulence. rPly4 activated murine bone marrow-derived dendritic cells in a Toll-like receptor (TLR) 4-dependent manner. The rPly4 alone was able to produce hemolytic activity at high concertation and penetrate the lipid bilayer. We further demonstrated that domain 4 of Ply involved in the virulence of the bacteria in mouse model. In the absence of apoptotic activity, the virulence level caused by rPly4 was similar to that of full length Ply. Our data suggested that domain 4 of Ply alone with TLR4 agonist and hemolytic activity may play roles in virulence of Streptococcus pneumoniae.


Assuntos
Hemólise , Estreptolisinas/química , Estreptolisinas/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Humanos , Camundongos Endogâmicos ICR , Domínios Proteicos , Proteínas Recombinantes/farmacologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/farmacologia , Relação Estrutura-Atividade , Virulência/efeitos dos fármacos
12.
Proc Natl Acad Sci U S A ; 116(27): 13352-13357, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31209022

RESUMO

Pneumolysin (PLY), a major virulence factor of Streptococcus pneumoniae, perforates cholesterol-rich lipid membranes. PLY protomers oligomerize as rings on the membrane and then undergo a structural transition that triggers the formation of membrane pores. Structures of PLY rings in prepore and pore conformations define the beginning and end of this transition, but the detailed mechanism of pore formation remains unclear. With atomistic and coarse-grained molecular dynamics simulations, we resolve key steps during PLY pore formation. Our simulations confirm critical PLY membrane-binding sites identified previously by mutagenesis. The transmembrane ß-hairpins of the PLY pore conformation are stable only for oligomers, forming a curtain-like membrane-spanning ß-sheet. Its hydrophilic inner face draws water into the protein-lipid interface, forcing lipids to recede. For PLY rings, this zone of lipid clearance expands into a cylindrical membrane pore. The lipid plug caught inside the PLY ring can escape by lipid efflux via the lower leaflet. If this path is too slow or blocked, the pore opens by membrane buckling, driven by the line tension acting on the detached rim of the lipid plug. Interestingly, PLY rings are just wide enough for the plug to buckle spontaneously in mammalian membranes. In a survey of electron cryo-microscopy (cryo-EM) and atomic force microscopy images, we identify key intermediates along both the efflux and buckling pathways to pore formation, as seen in the simulations.


Assuntos
Membrana Celular/efeitos dos fármacos , Estreptolisinas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Membrana Celular/metabolismo , Colesterol/metabolismo , Microscopia Crioeletrônica , Bicamadas Lipídicas/metabolismo , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Estreptolisinas/farmacologia
13.
Cell Microbiol ; 21(9): e13064, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31155820

RESUMO

Mast cells are implicated in the innate proinflammatory immune defence against bacterial insult, but the mechanisms through which mast cells respond to bacterial encounter are poorly defined. Here, we addressed this issue and show that mast cells respond vividly to wild type Streptococcus equi by up-regulating a panel of proinflammatory genes and by secreting proinflammatory cytokines. However, this response was completely abrogated when the bacteria lacked expression of sagA, whereas the lack of a range of other potential virulence genes (seeH, seeI, seeL, seeM, hasA, seM, aroB, pyrC, and recA) had no effect on the amplitude of the mast cell responses. The sagA gene encodes streptolysin S, a lytic toxin, and we next showed that the wild type strain but not a sagA-deficient mutant induced lysis of mast cells. To investigate whether host cell membrane perturbation per se could play a role in the activation of the proinflammatory response, we evaluated the effects of detergent- and pneumolysin-dependent lysis on mast cells. Indeed, exposure of mast cells to sublytic concentrations of all these agents resulted in cytokine responses of similar amplitudes as those caused by wild type streptococci. This suggests that sublytic membrane perturbation is sufficient to trigger full-blown proinflammatory signalling in mast cells. Subsequent analysis showed that the p38 and Erk1/2 signalling pathways had central roles in the proinflammatory response of mast cells challenged by either sagA-expressing streptococci or detergent. Altogether, these findings suggest that sagA-dependent mast cell membrane perturbation is a mechanism capable of activating the innate immune response upon bacterial challenge.


Assuntos
Proteínas de Bactérias/metabolismo , Inflamação/metabolismo , Mastócitos/imunologia , Streptococcus equi/genética , Streptococcus equi/patogenicidade , Estreptolisinas/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Citocinas/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/genética , Estreptolisinas/genética , Estreptolisinas/farmacologia , Virulência/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Neuroinflammation ; 16(1): 105, 2019 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-31103037

RESUMO

BACKGROUND: Streptococcus pneumoniae is the cause of a highly lethal form of meningitis in humans. Microglial cells in the brain represent the first line of defense against pathogens, and they participate in the inflammatory response. The cholesterol-dependent cytolysin pneumolysin and the bacterial capsule are key pathogenic factors, known to exacerbate the course of pneumococcal meningitis. METHODS: We utilized live imaging and immunostaining of glial cells in dissociated and acute brain slice cultures to study the effect of pneumococcal factors, including the cholesterol-dependent cytolysin pneumolysin and the pneumococcal capsule, on microglial motility and taxis. RESULTS: In brain tissue, primary microglia cells showed an enhanced response towards lysates from bacteria lacking capsules and pneumolysin as they moved rapidly to areas with an abundance of bacterial factors. The presence of bacterial capsules and pneumolysin cumulatively inhibited microglial taxis. In mixed cultures of astrocytes and microglia, the motility of microglia was inhibited by capsular components within minutes after exposure. The reduced motility was partially reversed by mannan, a mannose receptor inhibitor. The effects on microglia were not mediated by astrocytes because pure microglial cells responded to various pneumococcal lysates similarly with distinct cell shape changes as seen in mixed cultures. CONCLUSIONS: Our data indicate that microglia possess the capacity for a very agile response towards bacterial pathogens, but key pathogenic factors, such as pneumococcal capsules and pneumolysin, inhibited this response shortly after a bacterial challenge. Furthermore, we demonstrate for the first time that the bacterial capsule affects cellular behaviors such as motility and taxis.


Assuntos
Cápsulas Bacterianas/fisiologia , Movimento Celular/fisiologia , Quimiotaxia/fisiologia , Microglia/fisiologia , Streptococcus pneumoniae/fisiologia , Estreptolisinas/fisiologia , Animais , Proteínas de Bactérias/farmacologia , Proteínas de Bactérias/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/microbiologia , Técnicas de Cultura de Órgãos , Estreptolisinas/farmacologia
15.
Sci Rep ; 9(1): 5371, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926881

RESUMO

Group A Streptococcus (GAS) infection is associated with a variety of human diseases. Previous studies indicate GAS infection leads to RAW264.7 cell death, but the mechanism is unclear. Here, analyzing the timing of reactive oxygen species (ROS) production and using mitochondrial ROS scavenger, we found the wild type GAS-induced RAW264.7 cell death was associated with mitochondrial ROS. The wild type GAS infection could activate glycogen synthase kinase-3ß (GSK-3ß). Inhibition of GSK-3ß activity by lithium chloride or decreasing GSK-3ß expression by lentivirus-mediated short hairpin RNA for GSK-3ß could not only decrease the wild type GAS-induced mitochondrial ROS generation, mitochondria damage and cell death, but also reduced GAS intracellular replication. Streptolysin S (SLS), a GAS toxin, played the important role on GAS-induced macrophage death. Compared to the wild type GAS with its isogenic sagB mutant (SLS mutant)-infected macrophages, we found sagB mutant infection caused less mitochondrial ROS generation and cell death than those of the wild type GAS-infected ones. Furthermore, the sagB mutant, but not the wild type or the sagB-complementary mutant, could induce GSK-3ß degradation via a proteasome-dependent pathway. These results suggest that a new mechanism of SLS-induced macrophage death was through inhibiting GSK-3ß degradation and further enhancing mitochondrial damage.


Assuntos
Proteínas de Bactérias/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Macrófagos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/farmacologia , Animais , Camundongos , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Infecções Estreptocócicas/enzimologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/enzimologia
16.
Mol Med Rep ; 19(1): 414-422, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30431141

RESUMO

Streptococcus pyogenes (GAS) is a clinically significant bacterial strain that causes bacterial arthritis, osteomyelitis and implant infections. Infection complications can lead to serious bone destruction. Osteoclasts, the only type of cell with bone resorption function, participate in this process. Streptolysin O (SLO) is produced by almost all clinical Streptococcus pyogenes isolates. However, the role of SLO in bone infection caused by GAS had not been previously examined. The current study was performed to define the effects of SLO on receptor activator of NF­κB ligand­stimulated osteoclast differentiation in vitro. Results demonstrated that SLO decreased the phosphorylation of p65 and NF­κB inhibitor α, suppressed c­FOS and nuclear factor of activated T­cells cytoplasmic 1, and downregulated the expression of osteoclast marker genes. SLO also induced apoptosis of mature osteoclasts. The results suggested that SLO blocked osteoclast activation during GAS infection. These findings may prove useful in the development of novel strategies for treating GAS­associated bone infectious diseases.


Assuntos
NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ligante RANK/metabolismo , Transdução de Sinais/efeitos dos fármacos , Streptococcus pyogenes/metabolismo , Estreptolisinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Biomarcadores/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Camundongos , Osteoclastos/metabolismo , Células RAW 264.7
17.
FASEB J ; 33(1): 275-285, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29979630

RESUMO

Bacterial pore-forming toxins compromise plasmalemmal integrity, leading to Ca2+ influx, leakage of the cytoplasm, and cell death. Such lesions can be repaired by microvesicular shedding or by the endocytic uptake of the injured membrane sites. Cells have at their disposal an entire toolbox of repair proteins for the identification and elimination of membrane lesions. Sphingomyelinases catalyze the breakdown of sphingomyelin into ceramide and phosphocholine. Sphingomyelin is predominantly localized in the outer leaflet, where it is hydrolyzed by acid sphingomyelinase (ASM) after lysosomal fusion with the plasma membrane. The magnesium-dependent neutral sphingomyelinase (NSM)-2 is found at the inner leaflet of the plasmalemma. Because either sphingomyelinase has been ascribed a role in the cellular stress response, we investigated their role in plasma membrane repair and cellular survival after treatment with the pore-forming toxins listeriolysin O (LLO) or pneumolysin (PLY). Jurkat T cells, in which ASM or NSM-2 was down-regulated [ASM knockdown (KD) or NSM-2 KD cells], showed inverse reactions to toxin-induced membrane damage: ASM KD cells displayed reduced toxin resistance, decreased viability, and defects in membrane repair. In contrast, the down-regulation of NSM-2 led to an increase in viability and enhanced plasmalemmal repair. Yet, in addition to the increased plasmalemmal repair, the enhanced toxin resistance of NSM-2 KD cells also appeared to be dependent on the activation of p38/MAPK, which was constitutively activated, whereas in ASM KD cells, the p38/MAPK activation was constitutively blunted.-Schoenauer, R., Larpin, Y., Babiychuk, E. B., Drücker, P., Babiychuk, V. S., Avota, E., Schneider-Schaulies, S., Schumacher, F., Kleuser, B., Köffel, R., Draeger, A. Down-regulation of acid sphingomyelinase and neutral sphingomyelinase-2 inversely determines the cellular resistance to plasmalemmal injury by pore-forming toxins.


Assuntos
Toxinas Bacterianas/farmacologia , Membrana Celular/metabolismo , Proteínas de Choque Térmico/farmacologia , Proteínas Hemolisinas/farmacologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Estreptolisinas/farmacologia , Proteínas de Bactérias/farmacologia , Transporte Biológico , Sistemas CRISPR-Cas , Cálcio/metabolismo , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Humanos , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Sci Rep ; 8(1): 15846, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30367146

RESUMO

The lytic capacity of cholesterol-dependent cytolysins is enhanced in the extracellular calcium-free environment through a combination of limited membrane repair and diminished membrane toxin removal. For a typical neurotoxin of the group, pneumolysin, this effect has already been observed at reduced (1 mM) calcium conditions, which are pathophysiologically relevant. Here, we tested another neurotoxin of the group, listeriolysin O from L. monocytogenes, active in the primary vacuole after bacterium phagocytosis in host cells. Reduced calcium did not increase the lytic capacity of listeriolysin (in contrast to pneumolysin), while calcium-free conditions elevated it 2.5 times compared to 10 times for pneumolysin (at equivalent hemolytic capacities). To clarify these differences, we analyzed membrane vesicle shedding, known to be a calcium-dependent process for toxin removal from eukaryotic cell membranes. Both pneumolysin and listeriolysin initiated vesicle shedding, which was completely blocked by the lack of extracellular calcium. Lack of calcium, however, elevated the toxin load per a cell only for pneumolysin and not for listeriolysin. This result indicates that vesicle shedding does not play a role in the membrane removal of listeriolysin and outlines a major difference between it and other members of the CDC group. Furthermore, it provides new tools for studying membrane vesicle shedding.


Assuntos
Toxinas Bacterianas/farmacologia , Cálcio/química , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proteínas de Choque Térmico/farmacologia , Proteínas Hemolisinas/farmacologia , Listeria monocytogenes/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Proteínas de Bactérias/farmacologia , Células Cultivadas , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estreptolisinas/farmacologia
19.
Anim Reprod Sci ; 196: 130-137, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30033189

RESUMO

The ICSI-sperm mediated gene transfer (ICSI-SMGT) has been used to produce transgenic mice with high efficiency; however, the efficiency of this technique in farm animals is still less than desirable. Pretreatment of sperm with membrane destabilizing agents can improve the efficiency of ICSI in cattle. The objective of the present study was to evaluate streptolysin-O (SLO) as a novel treatment to permeabilize the bovine sperm membrane and assess its effect on efficiency of generating transgenic embryos by ICSI-SMGT. First, there was evaluation of the plasma membrane integrity (SYBR/PI), acrosome membrane integrity (PNA/FITC), DNA damage (TUNEL) and binding capacity of exogenous DNA (Nick Translation) in bull sperm treated with SLO. Subsequently, there was assessment of embryonic development and the efficiency in generating transgenic embryos with enhanced expression of the gene for green fluorescent protein (EGFP). Results indicate that SLO efficiently permeabilizes the plasma and acrosome membranes of bull spermatozoa and increases binding of exogenous DNA mostly to the post-acrosomal region and tail without greatly affecting the integrity of the DNA. Furthermore, treatment of bull spermatozoa with SLO prior to the injection of oocytes by ICSI-SMGT significantly increased the rate of embryo expression of the EGFP gene. Future experiments are still needed to determine the effect of this treatment on the development and transgene expression in fetuses and animals produced by ICSI-SMGT.


Assuntos
Bovinos/embriologia , Técnicas de Transferência de Genes/veterinária , Proteínas de Fluorescência Verde/metabolismo , Injeções de Esperma Intracitoplásmicas/veterinária , Espermatozoides/fisiologia , Animais , Feminino , Masculino , Gravidez , Espermatozoides/efeitos dos fármacos , Estreptolisinas/farmacologia
20.
PLoS One ; 13(7): e0201530, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30059559

RESUMO

Pleural infection/empyema is common and its incidence continues to rise. Streptococcus pneumoniae is the commonest bacterial cause of empyema in children and among the commonest in adults. The mesothelium represents the first line of defense against invading microorganisms, but mesothelial cell responses to common empyema pathogens, including S. pneumoniae, have seldom been studied. We assessed mesothelial cell viability in vitro following exposure to common empyema pathogens. Clinical isolates of S. pneumoniae from 25 patients with invasive pneumococcal disease and three reference strains were tested. All potently induced death of cultured mesothelial cells (MeT-5A) in a dose- and time-dependent manner (>90% at 107 CFU/mL after 24 hours). No significant mesothelial cell killing was observed when cells were co-cultured with Staphylococcus aureus, Streptococcus sanguinis and Streptococcus milleri group bacteria. S. pneumoniae induced mesothelial cell death via secretory product(s) as cytotoxicity could be: i) reproduced using conditioned media derived from S. pneumoniae and ii) in transwell studies when the bacteria and mesothelial cells were separated. No excess cell death was seen when heat-killed S. pneumoniae were used. Pneumolysin, a cytolytic S. pneumoniae toxin, induced cell death in a time- and dose-dependent manner. S. pneumoniae lacking the pneumolysin gene (D39 ΔPLY strain) failed to kill mesothelial cells compared to wild type (D39) controls, confirming the necessity of pneumolysin in D39-induced mesothelial cell death. However, pneumolysin gene mutation in other S. pneumoniae strains (TIGR4, ST3 and ST23F) only partly abolished their cytotoxic effects, suggesting different strains may induce cell death via different mechanisms.


Assuntos
Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Pleura/microbiologia , Pleura/patologia , Streptococcus pneumoniae/patogenicidade , Proteínas de Bactérias/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Criança , Empiema Pleural/metabolismo , Empiema Pleural/microbiologia , Empiema Pleural/patologia , Células Epiteliais/patologia , Epitélio/microbiologia , Epitélio/patologia , Epitélio/fisiologia , Humanos , Infecções Pneumocócicas/metabolismo , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/patologia , Streptococcus pneumoniae/isolamento & purificação , Streptococcus pneumoniae/fisiologia , Estreptolisinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...